WO2018066585A1 - Anticorps monoclonal anti-fzd10, et application de celui-ci - Google Patents

Anticorps monoclonal anti-fzd10, et application de celui-ci Download PDF

Info

Publication number
WO2018066585A1
WO2018066585A1 PCT/JP2017/036081 JP2017036081W WO2018066585A1 WO 2018066585 A1 WO2018066585 A1 WO 2018066585A1 JP 2017036081 W JP2017036081 W JP 2017036081W WO 2018066585 A1 WO2018066585 A1 WO 2018066585A1
Authority
WO
WIPO (PCT)
Prior art keywords
fzd10
antibody
antigen
sample
binding fragment
Prior art date
Application number
PCT/JP2017/036081
Other languages
English (en)
Japanese (ja)
Inventor
陽介 原田
達己 横関
中村 祐輔
Original Assignee
オンコセラピー・サイエンス株式会社
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by オンコセラピー・サイエンス株式会社 filed Critical オンコセラピー・サイエンス株式会社
Priority to KR1020197012936A priority Critical patent/KR102531006B1/ko
Priority to US16/336,846 priority patent/US11079386B2/en
Priority to EP17858422.3A priority patent/EP3524677B1/fr
Priority to CA3038789A priority patent/CA3038789A1/fr
Priority to BR112019006422A priority patent/BR112019006422A2/pt
Priority to CN201780075513.0A priority patent/CN110036110B/zh
Priority to RU2019112825A priority patent/RU2765431C2/ru
Priority to JP2018543931A priority patent/JP7039039B2/ja
Publication of WO2018066585A1 publication Critical patent/WO2018066585A1/fr
Priority to JP2022004142A priority patent/JP2022068145A/ja

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • G01N33/534Production of labelled immunochemicals with radioactive label
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57492Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds localized on the membrane of tumor or cancer cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6872Intracellular protein regulatory factors and their receptors, e.g. including ion channels
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70596Molecules with a "CD"-designation not provided for elsewhere in G01N2333/705
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/71Assays involving receptors, cell surface antigens or cell surface determinants for growth factors; for growth regulators
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present invention relates to a monoclonal antibody against FZD10, a method for diagnosing FZD10-related diseases using the antibody, a method for detecting FZD10 protein, a method for determining the efficacy after treatment with an FZD10 inhibitor, and a high therapeutic effect with an FZD10 inhibitor
  • the present invention relates to a method for screening a subject and a diagnostic reagent containing the antibody.
  • Frizzled proteins are a family of G protein-coupled receptors that have binding sites for Wnt protein ligands. Genetic analysis has so far identified 18 Wnt genes and 10 Frizzled genes (FZD1 to FZD10), all of which are highly structurally similar. Frizzled protein is a seven-transmembrane protein with an extracellular cysteine-rich domain at the N-terminus. This cysteine-rich domain is the Wnt ligand binding site. The binding between the Wnt ligand and the Frizzled receptor is not necessarily one-to-one, and it has been confirmed that one type of Wnt ligand binds to multiple types of Frizzled receptors and one type of Frizzled receptor binds to multiple types of Wnt ligands.
  • Wnt signaling pathway is activated by the binding of Wnt ligand and Frizzled receptor.
  • Wnt signaling pathways There are several Wnt signaling pathways that activate the ⁇ -catenin pathway and several pathways that do not mediate ⁇ -catenin, and it is thought that different pathways are activated by combinations of Wnt ligands and Frizzled receptors. ing.
  • the Wnt / ⁇ -catenin signaling pathway activated by receptor binding is mediated by Dishevelled (Dsh), a cytoplasmic protein that interacts directly with the Frizzled receptor, to stabilize and accumulate ⁇ -catenin in the cytoplasm Bring.
  • Dsh Dishevelled
  • ⁇ -catenin localizes to a cytoplasmic degradation complex that includes the tumor suppressor proteins colon adenomatous polyposis (APC) and auxin. These proteins serve as critical scaffolds for glycogen synthase kinase (GSK) -3 ⁇ to bind to and phosphorylate ⁇ -catenin, which is designated for degradation via the ubiquitin / proteasome pathway.
  • APC tumor suppressor proteins colon adenomatous polyposis
  • auxin serve as critical scaffolds for glycogen synthase kinase (GSK) -3 ⁇ to bind to and phosphorylate ⁇ -catenin, which is designated for degradation via the ubiquitin / proteasome pathway.
  • Patent Document 1 ⁇ -catenin-independent pathways have been shown to be involved in a number of processes, but are involved in intracellular planar polarity (PCP), cell motility and adhesion involved in the control of the cytoskeletal system
  • PCP planar polarity
  • Wnt / Ca 2+ pathways involved in the control of myogenesis via protein kinase A, and the like.
  • the Frizzled receptor can be dimerized, and it has been reported that this dimerization is involved in the activation of the Wnt signal pathway (Non-patent Document 1).
  • FZD10 (reference sequence: Genbank accession number NM_007197.3 (SEQ ID NO: 21)) mRNA is found in many cancer cell lines, including cervical, gastrointestinal and glioblastoma cell lines, and approximately 40% There are reports that it is up-regulated in primary gastric cancer, primary colon cancer and most synovial sarcoma tissues (Patent Documents 1 and 2, Non-Patent Documents 2 and 3).
  • Diseases associated with overexpression of FZD10 protein include synovial sarcoma, colorectal cancer (colon cancer), gastric cancer, chronic myelogenous leukemia (CML), and acute myeloid leukemia (AML) (Patent Literature) 3-5).
  • FZD10 is considered to be a suitable target for anticancer drugs, and the growth of synovial sarcoma cells is suppressed by siRNA specific for FZD10, and the antibody against FZD10 is a mouse graft model of synovial sarcoma. Have antitumor activity (Patent Documents 3 and 4).
  • monoclonal antibodies against FZD10 can be expected to serve as diagnostic agents in therapy.
  • monoclonal antibodies such as trastuzumab diagnostics, rituximab diagnostics, and bevacizumab diagnostics for breast cancer, malignant lymphoma, and colon cancer
  • monoclonal antibodies against other molecular targets are under development and their diagnostic effects are under evaluation. These diagnostic agents are expected to lead to more effective treatments from the viewpoint of effective patient selection in therapeutic agents.
  • Diagnostic agents in the treatment of tumors with molecularly targeted therapeutics are important to detect cellular proteins that are overexpressed in the majority of the target tumor and are not expressed in normal tissue or minimally expressed .
  • it is difficult to specifically detect a protein expressed in a tumor with high sensitivity and it is also difficult to obtain an antibody against such a protein.
  • several antibodies are commercially available for FZD10, which is considered a target for anticancer drugs.
  • FZD10-expressing cells with a commercially available antibody obtained by the present inventors, a positive signal (false positive) was sometimes generated even in cells with a low expression level of FZD10.
  • an object of the present invention is to provide an antibody that binds to FZD10 specifically and with high sensitivity.
  • the present inventors searched for an antibody that specifically binds to FZD10 from monoclonal antibodies obtained by immunizing mice with FZD10 antigen, and the specific antibody specifically binds to recombinant human FZD10 protein.
  • the present invention relates to: [1] CDR1 comprising the amino acid sequence shown in SEQ ID NO: 1; CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 2; and CDR3 comprising the amino acid sequence shown in SEQ ID NO: 3
  • a heavy chain variable region comprising, and CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 4; CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 5; and CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6
  • the heavy chain variable region comprising the amino acid sequence represented by SEQ ID NO: 7 and the light chain variable region comprising the amino acid sequence represented by SEQ ID NO: 8, or both, An antibody or antigen-binding fragment thereof.
  • [3] The antibody or antigen-binding fragment thereof according to [1] or [2], which specifically recognizes a polypeptide consisting of the amino acid sequence represented by SEQ ID NO: 9.
  • [5] The antibody or antigen-binding fragment thereof according to any one of [1] to [4], which is bound to an affinity label, an enzyme label, a radioisotope label, or a fluorescent label.
  • [7] comprising the antibody or antigen-binding fragment thereof according to any one of [1] to [5], diagnosing an FZD10-related disease, determining efficacy after treatment with an FZD10 inhibitor, or therapeutic effect with an FZD10 inhibitor Reagents for screening high subjects.
  • a method for diagnosing an FZD10-related disease or a predisposition for developing the disease in a subject comprising the following steps: (a) contacting the sample isolated from the subject with the antibody or antigen-binding fragment thereof according to any one of [1] to [5]; (b) detecting FZD10 protein in the sample by detecting binding of the antibody or antigen-binding fragment thereof to the sample: and (c) comparing the FZD10 protein level in the sample to a control, wherein the subject is suffering from or at risk of developing the disease when the FZD10 protein level is high compared to the control. Stage.
  • the cancer is selected from the group consisting of synovial sarcoma, lung cancer, esophageal cancer, colorectal cancer (colon cancer), gastric cancer, chronic myeloid leukemia (CML) and acute myeloid leukemia (AML)
  • CML chronic myeloid leukemia
  • AML acute myeloid leukemia
  • a method for detecting FZD10 protein in a sample comprising the following steps: (a) contacting a sample isolated from a subject with the antibody or antigen-binding fragment thereof according to any one of [1] to [5]; and (b) detecting FZD10 protein in the sample by detecting binding of the antibody or antigen-binding fragment thereof to the sample.
  • a method for determining efficacy after treatment with an FZD10 inhibitor in a subject comprising the following steps: (a) contacting the sample isolated from the subject with the antibody or antigen-binding fragment thereof according to any one of [1] to [5]; (b) detecting FZD10 protein in the sample by detecting binding of the antibody or antigen-binding fragment thereof to the sample; (c) comparing the FZD10 protein level in the sample with the expression level before drug administration, indicating that the subject had a medicinal effect when the FZD10 protein level was low compared to before drug administration , Stage.
  • a method for screening a subject having a high therapeutic effect by an FZD10 inhibitor comprising the following steps: (a) contacting the sample isolated from the subject with the antibody or antigen-binding fragment thereof according to any one of [1] to [5]; (b) detecting FZD10 protein in the sample by detecting binding of the antibody or antigen-binding fragment to the sample; (c) comparing the FZD10 protein level in the sample with a control, wherein the FZD10 inhibitor has a higher therapeutic effect in the subject when the FZD10 protein level is comparable or higher than the control It is shown that the stage.
  • a method for producing an antibody capable of binding to FZD10 protein or a partial peptide thereof comprising the following steps: (a) culturing a cell containing a vector into which the polynucleotide according to [6] is introduced; and (b) recovering the antibody from the cell culture or culture medium.
  • the invention further relates to: [16] A method for detecting a diagnostic marker for an FZD10-related disease or a predisposition to develop the disease, comprising the following steps: (a) contacting a sample isolated from the subject with the antibody or antigen-binding fragment thereof according to any one of [1] to [5]; and (b) detecting FZD10 protein in the sample as the marker by detecting binding of the antibody or antigen-binding fragment to the sample. [17] The antibody or antigen-binding fragment thereof according to any one of [1] to [5] for use in diagnosis of an FZD10-related disease or a predisposition to developing the disease.
  • [18] Use of the antibody or antigen-binding fragment thereof according to any one of [1] to [5] in the manufacture of a reagent for diagnosing a FZD10-related disease or a predisposition to the disease.
  • a method for detecting a drug efficacy marker of an FZD10 inhibitor comprising the following steps: (a) contacting a sample isolated from the subject with the antibody or antigen-binding fragment thereof according to any one of [1] to [5]; and (b) detecting FZD10 protein in the sample as the marker by detecting binding of the antibody or antigen-binding fragment to the sample.
  • a method for detecting an FZD10 inhibitor treatment responsive marker comprising the following steps: (a) contacting a sample isolated from the subject with the antibody or antigen-binding fragment thereof according to any one of [1] to [5]; and (b) detecting FZD10 protein in the sample as the marker by detecting binding of the antibody or antigen-binding fragment to the sample.
  • FIG. 1 is a diagram showing the expression of FZD10 mRNA in each cancer cell line in Cancer Line Encyclopedia (CCLE), which is an mRNA expression analysis database using cell lines for each cancer type.
  • FIG. 2 is a photomicrograph showing the specificity of the anti-FZD10 antibody (10A8H4G4) in immunohistochemical staining using the FZD10 forced expression system cell line.
  • an anti-FZD10 antibody (10A8H4G4) specific staining was observed in a paraffin section prepared from the FZD10 forced expression cell line (FZD10 / DLD1), but an Empty vector was introduced as a negative control. It was not stained with the cell line (Mock / DLD1).
  • FZD10 (L164) Ab, FZD10PolyclonalAb
  • staining was observed in both FZD10 / DLD1 and Mock / DLD1 as a result of immunohistochemical staining.
  • the specificity of the anti-FZD10 antibody (10A8H4G4) was revealed compared with the commercially available antibody.
  • the graph below the photo shows the ratio (%) of FZD10 positive cells in the number of forced expression cells based on the results of immunohistochemical staining.
  • FIG. 3 shows the correlation between expression in flow cytometry and immunohistochemical staining using various cell lines with different expression levels of FZD10.
  • A A histogram showing the results of flow cytometry using 10A8H4G4 in which the expression of FZD10 on the cell membrane of each cell line was detected.
  • SYO-1, T.T, and H727 were FZD10 expressing cell lines, and LoVo was a FZD10 non-expressing cell line.
  • B Photomicrograph showing the results of immunohistochemical staining using paraffin sections prepared from each cell line. In addition, the graph below the photograph shows the ratio (%) of FZD10-positive cells in each cell line calculated from the results of immunohistochemical staining. Staining was observed in FZD10-expressing cell lines such as SYO-1, T.T, and H727, but not in LoVo (non-expressing cell line).
  • FIG. 4 shows the correlation between immunohistochemical staining and expression in RealTime-PCR using paraffin sections prepared from mouse Xenograft tumors of SYO-1 and COLO201 cell lines and lung cancer clinical specimens 1-6.
  • the ratio of the number of FZD10-positive cells in the tumor cells was calculated from the results of immunohistochemical staining of B: A and shown in the graph.
  • C The results correlated with immunohistochemical staining were obtained by the expression analysis of the same case by RealTime-PCR. Analysis using clinical specimens revealed high specificity of the anti-FZD10 antibody (10A8H4G4) of the present invention in the immunohistochemical staining method.
  • the present invention provides an anti-FZD10 monoclonal antibody capable of specifically binding to FZD10 protein or a partial peptide thereof.
  • the present invention provides evidence that the anti-FZD10 monoclonal antibody of the present invention has high specificity in the detection of FZD10 protein in immunohistochemical staining methods.
  • the anti-FZD10 antibody (10A8H4G4) of the present invention has the following amino acid sequence in at least the variable region.
  • 10A8H4G4G4, light chain variable region amino acid sequence (excluding signal sequence): DIVMTQSPSSLTVTAGEKVTMSCKSSQSLLNSGNQKNYLTWYQQKPGQPPKLLIYWASTRKSGVPDRFTGSGSGTDFTLTISSVQAEDLAVYYCQNDYSYPVTFGAGTKLELKRAD SEQ ID NO: 8
  • the antibody of the present invention can also be produced by a recombinant technique using DNA
  • the antibody of the present invention was obtained by screening and selecting an antibody having binding ability to FZD10 by ELISA from a plurality of antibody-producing hybridomas obtained by immunization of mice.
  • the antibodies selected by ELISA were further selected by immunostaining.
  • Antibodies that show negative results were selected.
  • an antibody having a weak binding force is attached as a background.
  • the antibody of the present invention specifically binds to FZD10. Therefore, the antibody of the present invention is useful as a tool for detecting FZD10 or a cell or tissue expressing FZD10. Further, the antibody of the present invention can be used as a label bound to a label capable of detecting the antibody, and the label is, for example, a cancer cell or cancer expressing FZD10 such as colorectal cancer. It is more preferable in detecting the tissue.
  • the label that binds to the antibody of the present invention may be any label that can detect an antibody bound to FZD10, such as an affinity label (eg, biotin, avidin, etc.), an enzyme label (eg, horseradish peroxidase, alkaline phosphatase, etc.) And fluorescent labels (for example, FITC, rhodamine and the like).
  • an affinity label eg, biotin, avidin, etc.
  • an enzyme label eg, horseradish peroxidase, alkaline phosphatase, etc.
  • fluorescent labels for example, FITC, rhodamine and the like.
  • the antibody of the present invention when used as a diagnostic agent for selection of cancer treatment patients, the antibody of the present invention can be used as it is, but it can be made into a composition suitable for various usage forms.
  • an isolated or purified antibody refers to an antibody that is substantially free of other cellular material, such as carbohydrates, lipids, and other contaminating proteins, from the cell or tissue source from which the antibody is derived.
  • the antibodies of the invention are isolated or purified.
  • polypeptide and “protein” are used interchangeably herein and refer to a polymer of amino acid residues. This term applies to non-natural amino acid polymers containing one or more non-natural amino acid residues as well as natural amino acid polymers. Non-natural amino acids include amino acid analogs and amino acid mimetics.
  • polynucleotide oligonucleotide
  • nucleic acid refers to a polymer of nucleotides.
  • FZD10-related disease is a cancer that expresses FZD10.
  • cancer refers to cancers that express FZD10, preferably those that overexpress the FZD10 gene, such as synovial sarcoma, lung cancer, esophageal cancer, colon Examples include, but are not limited to, rectal cancer (colon cancer), gastric cancer, chronic myeloid leukemia (CML), and acute myeloid leukemia (AML).
  • rectal cancer colon cancer
  • CML chronic myeloid leukemia
  • AML acute myeloid leukemia
  • cancer cells expressing FZD10 from mRNA expression analysis database “Cancer Cell Cell Line Encyclopedia” (CCLE) using a cell line for each cancer type. Also in this application, remarkable up-regulation was confirmed in lung cancer, esophageal cancer, and colorectal cancer (colorectal cancer) (FIG. 1).
  • antibody is intended to include immunoglobulins and fragments thereof that have specific reactivity to the designated protein or peptide thereof.
  • Antibodies can include antibodies fused with other proteins or labels, and antibody fragments.
  • antibodies are used herein in the broadest sense, specifically formed from intact monoclonal antibodies, polyclonal antibodies, at least two intact antibodies as long as they exhibit the desired biological activity. Multispecific antibodies (eg, bispecific antibodies), and antibody fragments are covered.
  • Antibody refers to all classes (eg, IgA, IgD, IgE, IgG, and IgM).
  • antibody fragment is a portion of an intact antibody and generally includes one or more antigen binding or variable regions of the intact antibody.
  • antibody fragments can include one or more antigen binding portions of an intact antibody.
  • the term “antigen-binding portion” or “antigen-binding fragment” of an antibody refers to one or more immunology of an antibody that retains the ability to specifically bind to an antigen (eg, FZD10). Active fragment. It has been shown that the antigen binding function of an antibody can be performed by fragments of a full-length antibody. Examples of antibody fragments include Fab, Fab ′, F (ab ′) 2 , and Fv fragments, linear antibodies, and single chain antibody molecules.
  • an antibody fragment binds with the same antigen that is recognized by the intact antibody.
  • antibody fragment also refers to a synthetic or genetically engineered polypeptide that binds to a specific antigen, eg, a polypeptide comprising a light chain variable region, an “Fv” fragment comprising heavy and light chain variable regions, a light chain It also includes a recombinant single-chain polypeptide molecule ("scFv protein") in which the variable regions of the chain and heavy chain are linked by a peptide linker, as well as a minimal recognition unit consisting of amino acid residues that mimic the hypervariable region.
  • scFv protein single-chain polypeptide molecule
  • an FZD10 protein and an antibody can be evaluated by, for example, competition between antibodies.
  • an antibody of the present invention for example, an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 7 and an antibody comprising a light chain variable region comprising the amino acid sequence of SEQ ID NO: 8 as the reference antibody.
  • the specificity of the candidate antibody can be evaluated.
  • a representative reference antibody is 10A8H4G4.
  • the amount of antibody that binds to FZD10 protein in the absence of the candidate antibody is 10%, 20%, 30%, or 40% when the reference antibody and FZD10 protein are reacted in the presence of the candidate antibody.
  • the binding of the reference antibody is inhibited by 50% or more, it can be determined that competition between the antibodies has occurred.
  • a partial peptide thereof can be used as long as the reference antibody binds.
  • a preferred partial peptide is the N-terminal extracellular domain peptide of FZD10 protein, for example, a partial peptide comprising the amino acid sequence of SEQ ID NO: 9.
  • an anti-FZD10 monoclonal antibody is used. Such antibodies are provided by methods well known in the art. Exemplary antibody production techniques used by the present invention are described below.
  • Monoclonal antibodies are obtained from a population of substantially homogeneous antibodies. That is, the individual antibodies that make up the population are identical except for natural mutations that may be present in minor amounts. Thus, the modifier “monoclonal” indicates the character of the antibody as not being a mixture with separate antibodies. For example, monoclonal antibodies can be produced using the hybridoma method first described by Kohler et al., Nature, 256: 495 (1975), or can also be produced by recombinant DNA methods (US 4,816,567). sell.
  • mice or other suitable host animals can be immunized with FZD10 polypeptide (FZD10 protein or a partial polypeptide thereof) to produce or produce antibodies that specifically bind to FZD10 polypeptide.
  • FZD10 polypeptide FZD10 protein or a partial polypeptide thereof
  • lymphocytes may be immunized with FZD10 polypeptide in vitro.
  • the lymphocytes are then fused with myeloma cells using a suitable fusing agent such as polyethylene glycol to form hybridoma cells (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)) .
  • the prepared hybridoma cells are seeded in an appropriate culture medium that preferably contains one or more substances that inhibit the growth or survival of unfused parent myeloma cells, and grown in that medium.
  • an appropriate culture medium that preferably contains one or more substances that inhibit the growth or survival of unfused parent myeloma cells, and grown in that medium.
  • the culture medium for hybridomas can typically include hypoxanthine, aminopterin, and thymidine. (HAT medium), these substances interfere with the growth of HGPRT-deficient cells.
  • Preferred myeloma cells are those that fuse efficiently, assist stable high level antibody production by selected antibody-producing cells, and are sensitive to a medium such as HAT medium.
  • Preferred myeloma cell lines include mouse myeloma lines such as those derived from MOPC-21 and MPC-11 mouse tumors available from Salk Institute Cell Distribution Center, San Diego, California USA, and American Type Culture Collection, Manassas , SP-2 cells or X63-Ag8-653 cells available from Virginia, USA.
  • Human myeloma cell lines and mouse-human heteromyeloma cell lines have also been described for the production of human monoclonal antibodies (Kozbor, J. Immunol., 133: 300 1 (1984); Brodeur et al., OcMonoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987)).
  • Culture medium in which hybridoma cells are growing is analyzed for production of monoclonal antibodies directed against the antigen.
  • the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by in vitro binding assays such as radioimmunoassay (RIA) or enzyme-linked immunosorbent assay (ELISA).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunosorbent assay
  • the binding affinity of a monoclonal antibody can be determined, for example, by the 30 Scatchard analysis of Munson et al., Anal. Biochem., 107: 220 (1980).
  • hybridoma cells After identifying hybridoma cells producing antibodies of the desired specificity, affinity, and / or activity, the clones can be subcloned by limiting dilution and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)). Examples of culture media suitable for this purpose include D-MEM medium or RPMI-1640 medium. In addition, hybridoma cells can be grown in vivo, such as ascites tumors in animals.
  • the monoclonal antibody secreted by the subclone may be purified to homogeneity.
  • separation and purification of antibodies can be performed according to separation and purification methods used for general proteins. For example, but not limited to, appropriately selecting and combining the use of column chromatography such as affinity chromatography, filters, ultrafiltration, salting out, dialysis, SDS polyacrylamide gel electrophoresis, and isoelectric focusing
  • column chromatography such as affinity chromatography, filters, ultrafiltration, salting out, dialysis, SDS polyacrylamide gel electrophoresis, and isoelectric focusing
  • proteins can be appropriately separated and isolated from culture medium, ascites fluid, or serum (Antibodies: A Laboratory Manual. Ed Harlow and David Lane, Cold Spring Harbor Laboratory (1988)).
  • Protein A and protein G columns can be used as affinity columns.
  • Exemplary protein A columns to be used include, for example, Hyper®D, POROS, and Sepharose® F.F. (Pharmacia).
  • Exemplary chromatography includes, in addition to affinity chromatography, for example, ion exchange chromatography, hydrophobic chromatography, gel filtration, reverse phase chromatography, adsorption chromatography, etc. (Strategies for Protein Purification and Characterization: A Laboratory Course Manual. Ed Daniel R. Marshak et al., Cold Spring Harbor Laboratory Press (1996)). Chromatographic procedures can be performed by liquid phase chromatography such as HPLC and FPLC.
  • the DNA encoding the monoclonal antibody is readily isolated and sequenced using conventional procedures (eg, by using oligonucleotide probes that can specifically bind to the genes encoding the heavy and light chains of the murine antibody). It is determined. Hybridoma cells serve as a preferred source of such DNA. Once the DNA is isolated, it is placed in an expression vector which is then E. coli cells, monkey COS cells, Chinese hamster ovary (CHO) cells that do not otherwise produce immunoglobulin proteins, or Transfection into host cells such as myeloma cells can achieve the synthesis of monoclonal antibodies in recombinant host cells. Review articles on recombinant expression in bacteria of DNA encoding the antibody include Skerra et al., Curr. Opinion in Immunol., 5: 256-262 (1993) and Pluckthun, Immunol. Revs., 130: 151- 188 (1992) is included.
  • Another method for producing a specific antibody or antibody fragment that is reactive to FZD10 is an expression library encoding an immunoglobulin gene or a portion thereof expressed in bacteria, FZD10 protein or its Screening with partial peptides.
  • complete Fab fragments, VH regions, and Fv regions can be expressed in bacteria using phage expression libraries. See, for example, Ward et al., Nature 341: 544-546 (1989); Huse et al., Science 246: 1275-1281 (1989); and McCafferty et al., Nature 348: 552-554 (1990).
  • I want For example, by screening such a library using the FZD10 peptide, immunoglobulin fragments reactive to FZD10 can be identified.
  • SCID-hu mice available from Genpharm) may be used to produce antibodies or fragments thereof.
  • the antibody or antibody fragment can be isolated from an antibody phage library produced using the techniques described in McCafferty et al., Nature, 348: 552-554 (1990). Clackson et al., Nature, 352: 624-628 (1991) and Marks et al., J MoL BioL, 222: 581-597 (1991), on isolation of mouse and human antibodies using phage libraries Each is listed.
  • the present invention provides antibodies suitable for diagnosis of FZD10-related diseases, determination of drug efficacy after treatment with FZD10 inhibitors, and screening of subjects with high therapeutic effects with FZD10 inhibitors.
  • the present invention has succeeded in establishing a mouse monoclonal antibody clone (10A8H4G4) capable of detecting FZD10 protein with high specificity in immunohistochemical staining and flow cytometry.
  • this antibody clone is used for immunohistochemical staining of lung cancer clinical specimens, positive findings are observed in lung cancer specimens that have been confirmed to have high FZD10 expression levels by RealTime-PCR, but low expression levels of FZD10 have been confirmed. Lung cancer specimens proved to be hardly stained.
  • the amino acid sequences of the heavy chain variable region (H chain V region) and light chain variable region (L chain V region) of the anti-FZD10 mouse monoclonal antibody clone (10A8H4G4) of the present invention are shown in SEQ ID NO: 7 and 8, respectively. ing.
  • the CDR (complementarity determining region) contained in the heavy chain variable region and the light chain variable region can be determined according to a method well known in the art. For example, by Kabat et al. (Kabat E. A. et al. (1991) Sequence of Proteins of Immunological Interest. 5th Edition) or Chothia et al. (Chothia et al. J. Mol. The described method is commonly used for CDR determination.
  • the CDR1, 2, and 3 of the heavy chain variable region of the anti-FZD10 mouse monoclonal antibody clone (10A8H4G4) of the present invention determined by Kabat definition are shown in SEQ ID NO: 1, 2, and 3, respectively.
  • the CDRs 1, 2, and 3 of the light chain variable region are shown in SEQ ID NO: 4, 5, and 6, respectively.
  • the present invention provides an antibody or antigen-binding fragment thereof capable of binding to FZD10 protein or a partial peptide thereof, comprising either or both of a heavy chain variable region and a light chain variable region, wherein the heavy chain variable
  • the area is CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 1; CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 2; and CDR3 comprising the amino acid sequence shown in SEQ ID NO: 3
  • the light chain variable region comprises CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 4; CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 5; and CDR3 comprising the amino acid sequence shown in SEQ ID NO: 6
  • An antibody or antigen-binding fragment thereof is provided.
  • the partial peptide of FZD10 protein to which the antibody of the present invention binds is preferably the N-terminal extracellular domain peptide of FZD10 protein, for example, corresponding to positions 21-161 of FZD10 protein (SEQ ID NO: 22) It comprises an amino acid sequence (SEQ ID NO: 9) and consists of an amino acid sequence selected from SEQ ID NO: 22. More preferably, the partial peptide of FZD10 protein in the present invention can consist of the amino acid sequence of SEQ ID NO: 9.
  • One example of a variable region is a heavy chain variable region comprising the amino acid sequence shown in SEQ ID NO: 7.
  • One example of a variable region is a light chain variable region comprising the amino acid sequence shown in SEQ ID NO: 8.
  • the invention includes either or both of a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 7 and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 8
  • a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 7
  • a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 8
  • the antibody of the present invention can be prepared by a conventional method.
  • an antibody can be prepared by incorporating a polynucleotide encoding an antibody polypeptide into an appropriate vector, introducing the vector into a host, and producing the antibody from the host.
  • Vandamme, A. M. et al., Eur. J. Biochem. (1990) 192, 767-75 See, for example, Vandamme, A. M. et al., Eur. J. Biochem. (1990) 192, 767-75).
  • the nucleic acid sequence of the polynucleotide encoding the variable region (V region) of the antibody of the present invention can be deduced from the amino acid sequence of the V region of the antibody of the present invention.
  • the nucleic acid sequences encoding the heavy chain variable region (VH) and light chain variable region (VL) of the antibody clone of the present invention for example, the nucleic acid sequences shown in SEQ ID NO: 10 and 11 can be used, respectively. .
  • Polynucleotide encoding the V region of the antibody of the present invention is a solid phase technology (Beaucage SL & Iyer RP, Tetrahedron (1992) 48, 2223-311; Matthes et al., EMBO J (1984) 3, 801-5) And can be synthesized based on the sequence information by a conventional method such as oligonucleotide synthesis technology (Jones et al. Nature (1986) 321, 522-5).
  • a polynucleotide encoding the antibody V region is incorporated into an expression vector comprising a polynucleotide encoding the antibody constant (C) region.
  • a polynucleotide encoding the antibody is incorporated into an expression vector so that the antibody gene can be expressed under the control of an expression control element (eg, enhancer, promoter).
  • An expression vector is used to transform host cells to express the antibody.
  • the polynucleotide encoding the antibody H chain and the polynucleotide encoding the L chain may be incorporated into separate expression vectors, and then the resulting recombinant expression vector is cotransformed into a host cell.
  • both the polynucleotide encoding the antibody H chain and the polynucleotide encoding the L chain may be incorporated together into a single expression vector, and then the resulting recombinant expression vector is transformed into a host cell ( For example, International Publication No. 94/11523).
  • the antibody gene can be expressed by a known method.
  • a conventional useful promoter for expression in mammalian cells, a conventional useful promoter, the expressed antibody gene, and a poly (A) signal (located downstream from the 3 ′ end of the antibody gene) can be operably linked.
  • a human cytomegalovirus immediate early promoter / enhancer system can be used as a useful promoter / enhancer system.
  • promoter / enhancer systems such as those derived from viruses (eg, retroviruses, polyomaviruses, adenoviruses and simian virus 40 (SV40)), and those derived from mammalian cells (eg, human elongation factor 1 ⁇ ( HEF1 ⁇ )) can also be used for the expression of antibodies in the present invention.
  • viruses eg, retroviruses, polyomaviruses, adenoviruses and simian virus 40 (SV40)
  • HEF1 ⁇ human elongation factor 1 ⁇
  • gene expression can be easily performed by the method of Mulligan et al. (Nature (1979) 277, 108-14.).
  • HEF1 ⁇ promoter / enhancer system gene expression can be easily performed by the method of Mizushima et al. (Nucleic Acids Res. (1990) 18, 5322.).
  • a conventional useful promoter a signal sequence for secreting the antibody of interest, and an antibody gene can be operably linked.
  • the promoter lacZ promoter or araB promoter can be used.
  • gene expression can be performed by the method of Ward et al. (Nature (1098) 341, 544-6 .; FASBE J. (1992) 6, 2422-7.), while the araB promoter. Is used, gene expression can be performed by the method of Better et al. (Science (1988) 240, 1041-3.).
  • the pelB signal sequence (Lei, SP et al, J. Bacteriol. (1987) 169, 4379-83.) Can be used.
  • the antibody secreted into the periplasmic space is isolated and then refolded so that the antibody assumes the proper conformation.
  • a replication origin derived from a virus eg, SV40, polyoma virus, adenovirus, bovine papilloma virus (BPV)
  • a virus eg, SV40, polyoma virus, adenovirus, bovine papilloma virus (BPV)
  • expression vectors include aminoglycoside phosphotransferase (APH) gene, thymidine kinase (TK) gene, E. coli xanthine-guanine phosphoribosyltransferase (Ecogpt) gene and dihydrofolate reductase (dhfr )
  • a selection marker gene such as a gene may be further included.
  • any expression system can be used, including eukaryotic and prokaryotic cell systems.
  • Eukaryotic cells include established cell lines of animals (eg, mammals, insects, molds and fungi, yeast).
  • Prokaryotic cells include bacterial cells such as E. coli cells.
  • the antibody used in the present invention is preferably expressed in mammalian cells such as CHO cells, COS cells, myeloma cells, BHK cells, Vero cells, and HeLa cells.
  • the transformed host cell is cultured in vitro or in vivo to produce the antibody of interest.
  • the host cell can be cultured by any known method.
  • the culture medium that can be used herein may be DMEM, MEM, RPMI-1640 or IMDM medium.
  • the culture medium may contain a serum supplement such as fetal calf serum (FCS).
  • transgenic animals can also be used as hosts for the production of recombinant antibodies.
  • a fusion gene is prepared by inserting an antibody gene into a predetermined site of a gene encoding a protein (eg, ⁇ -casein) originally produced in animal breast milk.
  • a DNA fragment containing the fusion gene into which the antibody gene has been introduced is injected into a non-human animal embryo, which is then introduced into a female animal.
  • the female animal having the embryo in its body produces a transgenic non-human animal.
  • the antibody of interest is secreted into the breast milk from the transgenic non-human animal or its progeny.
  • an appropriate hormone can be administered to the transgenic animal (Ebert, K.M. et al, Bio / Technology (1994) 12, 699-702).
  • the antibody expressed and produced as described above can be isolated from the body of the cell or host animal and purified. Isolation and purification of the antibody used in the present invention can be performed on an affinity column. Other methods conventionally used for antibody isolation and purification can also be used; therefore, the method is not particularly limited. For example, a variety of chromatography, filtration, ultrafiltration, salting out and dialysis can be used alone or in combination to isolate and purify the antibody of interest (Antibodies A Laboratory Manual. Ed. Harlow, David Lane, Cold Spring Harbor Laboratory, atory1988).
  • Antibody fragments Various techniques have been developed for the production of antibody fragments. Previously, these fragments were obtained through proteolytic digestion of intact antibodies (see, for example, Morimoto et al., Journal of Biochemical and Biophysical Methods 24: 107-117 (1992) and Brennan et al., Science , 229: 81 (1985)). However, these fragments can now be produced directly by recombinant host cells. For example, antibody fragments can be isolated from antibody phage libraries. Alternatively, Fab′-SH fragments may be recovered directly from E. coli and chemically coupled to form F (ab ′) 2 fragments (Carter et al., Bio / Technology 10: 163-167 ( 1992)).
  • F (ab ′) 2 fragments can be isolated directly from recombinant host cell culture.
  • the optimal antibody is a single chain Fv fragment (scFv). See WO 93/16185; US 5,571,894; and US 5,587,458.
  • the antibody fragment may also be a “linear antibody” as described in, for example, US Pat. No. 5,641,870. Such linear antibody fragments can have monospecificity or bispecificity.
  • the antibody of the present invention is optionally conjugated with an affinity label, enzyme label, radioisotope label, fluorescent label, or chemiluminescent label.
  • an affinity label for example, the presence of a detectable label that is present in a cancer tissue that expresses FZD10 makes it possible to determine the presence or absence of a cancer or tumor in the subject being diagnosed. Further, the spread of the disease can be determined by the localization of the label in the cancer tissue.
  • Suitable labels for use include, for example, fluorescent labels such as fluorescein and rhodamine; and enzyme labels such as luciferase.
  • the detectable / detectable label used is selected according to the imaging modality used.
  • conjugates of label and antibody can be made using protocols and techniques known in the art.
  • the antibody of the present invention may be bound to a desired label immediately before use, or may be provided as an antibody bound to the label.
  • Conjugates of antibody and label are N-succinimidyl-3- (2-pyridylditriol) propionate (SPDP), succinimidyl-4- (N-maleimidomethyl) cyclohexane-1-carboxylate, iminothiolane (IT), imide Bifunctional derivatives of esters (eg dimethyl adipate HCL), active esters (eg disuccinimidyl suberate), aldehydes (eg glutaraldehyde), bis-azide compounds (eg bis (p-azidobenzoyl) hexanediamine), Bis-diazonium derivatives (eg bis- (p-diazoniumbenzoyl) -ethylenediamine), diisocyanates (eg toluene 2,6-diisocyanate), and bis-active fluorine compounds (eg 1,5-difluoro-2,4-dinitrobenzene)
  • SPDP N-succ
  • a fusion protein comprising the antibody and a label can be made, eg, by recombinant techniques or peptide synthesis.
  • a fusion protein include a fusion protein of a labeled protein such as ECFP, EYFP, or EGFP and an antibody.
  • FZD10 is useful as a diagnostic marker for FZD10-related diseases and as a marker for evaluating the responsiveness to FZD10 inhibitors and the efficacy of the inhibitors in subjects suffering from the diseases. Therefore, the antibody of the present invention is a marker for diagnosing FZD10-related diseases such as cancer, for screening subjects with a high therapeutic effect by FZD10 inhibitors, or for determining the efficacy after treatment with FZD10 inhibitors. It can be used as a detection reagent.
  • the present invention provides a method for diagnosing a FZD10-related disease or a predisposition for developing the disease in a subject by detecting FZD10 protein using the antibody of the present invention in a sample isolated from the subject, and a therapeutic effect of an FZD10 inhibitor A method for screening a subject having a high incidence and a method for determining the efficacy after treatment with an FZD10 inhibitor are provided.
  • These methods include the following steps: (a) contacting a sample isolated from a subject with an antibody of the present invention or an antigen-binding fragment thereof; (b) detecting FZD10 protein in the sample by detecting binding of the antibody or antigen-binding fragment thereof to the sample: and (c) comparing the FZD10 protein level in the sample with a control.
  • the sample is a cell or tissue isolated from the subject, preferably a tissue isolated from the subject. Therefore, typically, each method of the invention is performed in vitro on a sample isolated from a subject.
  • Techniques for isolating tissues and cells from an object by techniques such as biopsy (biopsy) and blood collection are known.
  • a biological sample that has been removed from a subject through a therapeutic medical procedure such as a surgical operation
  • Cells and tissues isolated from the subject can be appropriately treated before contact with the antibody.
  • a tissue sample generally obtained from a subject can be sectioned after freezing and further fixed with alcohol, formalin, or the like to obtain a sample for immunohistological analysis.
  • a tissue sample, cultured cells or the like can be fixed with formalin and then embedded in paraffin to obtain a section for immunohistological analysis.
  • the binding of the antibody of the present invention or the antigen-binding fragment thereof to a sample can be detected by methods known to those skilled in the art. More specifically, after contacting the antibody of the present invention with the sample, those that did not bind to FZD10 in the sample are removed by washing, and then the antibody remaining in the sample is detected, thereby detecting the antibody of the present invention. And binding of FZD10 protein in the sample can be detected. At this time, when the antibody is directly labeled, the presence of the antibody of the present invention bound to the FZD10 protein can be detected by detecting the label.
  • the label is detectable such as an enzyme, a fluorescent substance, a luminescent substance, or particles
  • these labels can be detected immediately.
  • an affinity substance binding substance
  • biotin affinity label
  • the presence of the antibody can be captured by a binding partner such as labeled avidin.
  • the antibody of the present invention can be detected with a binding reagent for the antibody.
  • an antibody binding reagent an antibody against protein A or an antibody can be labeled and used to detect the antibody.
  • step (c) compared to the control level (normal control level, preferably the expression level of FZD10 protein in a sample isolated from a healthy subject not suffering from FZD10-related disease)
  • control level normal control level, preferably the expression level of FZD10 protein in a sample isolated from a healthy subject not suffering from FZD10-related disease
  • a high FZD10 protein level indicates that the subject is suffering from or at risk of developing a FZD10-related disease.
  • a control level preferably, an expression level of FZD10 protein in a tissue of a subject diagnosed with an FZD10-related disease.
  • the FZD10 protein level is comparable or higher, it indicates that the therapeutic effect of the FZD10 inhibitor in the subject is high.
  • a control level preferably, the expression level of FZD10 protein in a sample isolated from a subject before drug administration.
  • a patient who has been shown to have an FZD10-related disease by the diagnostic method of the present invention is likely to be treated with an FZD10 inhibitor. Therefore, following the diagnostic method of the present invention, an FZD10 inhibitor can be administered to a patient who has been shown to have an FZD10-related disease. Alternatively, the FZD10 inhibitor can be administered to a patient who has been shown to have a high therapeutic effect of the FZD10 inhibitor via screening. Furthermore, even if a patient who has been administered an FZD10 inhibitor is shown to have a therapeutic effect of the inhibitor, the FZD10 inhibitor can be subsequently administered to the same patient.
  • the present invention relates to a method for treating an FZD10-related disease, comprising the step of identifying any patient selected from the following group by the method of the present invention and administering an FZD10 inhibitor to the patient; A patient shown by the diagnostic method of the invention to have an FZD10-related disease; Patients who have been shown to be likely to benefit from FZD10 inhibitors; and Patients who have been treated with FZD10 inhibitors and have been shown to have therapeutic effects of the inhibitors.
  • a known compound can be used as the FZD10 inhibitor administered to a patient.
  • antibodies showing cytotoxic activity against FZD10-expressing cells and double-stranded RNA molecules that suppress FZD10 expression are also included in the FZD10 inhibitor.
  • examples of such antibodies and double-stranded RNA molecules include antibodies against FZD10 disclosed in WO2005 / 004912 or WO2007 / 148417, and siRNA specific for FZD10 disclosed in WO2006 / 013733. it can.
  • a control level measured from a biological sample known not to suffer from an FZD10-related disease is referred to as a “normal control level”. If the FZD10 protein level in the sample isolated from the subject is high compared to the normal control level, the subject can be diagnosed as having an FZD10-related disease to be treated.
  • a control level measured from a biological sample known to suffer from an FZD10-related disease eg, cancerous
  • a “disease control level eg, cancerous control level
  • the subject can be diagnosed as having a high therapeutic effect with the FZD10 inhibitor. Also, if the FZD10 protein level in a sample isolated from a subject after treatment with an FZD10 inhibitor is lower than the subject's disease control level prior to drug administration, the treatment was effective, i.e., the subject was treated with an FZD10 inhibitor. It can be diagnosed that the therapeutic effect is high.
  • normal cells (or tissues) obtained from an unaffected area (eg, non-cancerous area) of an organ having an FZD10-related disease (eg, cancer) to be treated are used as normal controls.
  • the control level is a result obtained by analyzing pre-measured FZD10 protein levels in a sample from a subject with known disease state (eg, cancerous or non-cancerous). Based on this, it may be determined by a statistical method.
  • the control level can be derived from a database of expression patterns from previously tested samples (cells or tissues). When the sample to be evaluated is a tissue sample, a sample derived from the same tissue is preferably used as the control sample.
  • the FZD10 protein level in a biological sample may be compared to a plurality of control levels measured from a plurality of reference samples. It is preferred to use a control level measured from a reference sample derived from a tissue type similar to the tissue type of the subject-derived biological sample. Furthermore, it is preferred to use a reference value for FZD10 protein level in a population with known disease state.
  • the reference value can be obtained by any method known in the art. For example, the range of the average value +/ ⁇ 2 S.D. or the average value +/ ⁇ 3 S.D. can be used as the reference value.
  • the FZD10 protein level in the sample is 10%, 25%, or 50% higher than the control level, or more than 1.1, 1.5, 2.0, 5.0, 10.0, or more If it is higher, it can be considered high.
  • the FZD10 protein level in the sample is, for example, 10%, 25%, or 50% lower than the control level, or greater than 1.1, 1.5, 2.0, 5.0, 10.0, or If it is lower, it can be regarded as low.
  • the FZD10-related disease is a cancer that expresses FZD10.
  • Cancers that express FZD10 are, for example, synovial sarcoma, lung cancer, esophageal cancer, colorectal cancer (colon cancer), gastric cancer, chronic myeloid leukemia (CML), and acute myeloid leukemia (AML) Although there is, it is not limited to these.
  • the present invention is a method for detecting a diagnostic marker for an FZD10-related disease or a predisposition to developing the disease, wherein the FZD10 protein in a sample is detected using the antibody of the present invention or an antigen-binding fragment thereof
  • a method comprising the step of detecting as FZD10 has been shown to have increased expression in certain cancer cells compared to normal tissues. Therefore, if the expression level of FZD10 can be specifically detected, it is useful as a diagnostic marker for the disease to which it is related.
  • a diagnostic marker for an FZD10-related disease or predisposition to the disease is FZD10 in a sample isolated from a subject detected by binding to an antibody of the invention or an antigen-binding fragment thereof.
  • the control level is a normal control level, preferably the expression level of FZD10 protein in a sample isolated from a healthy subject not suffering from an FZD10-related disease.
  • the control level is preferably the expression level in the same tissue as the tissue from which the cancer cells to be detected for the diagnostic marker are derived.
  • the invention also provides an antibody of the invention or an antigen-binding fragment thereof for use in diagnosing an FZD10-related disease or a predisposition to the disease.
  • the present invention provides the use of an antibody of the present invention or an antigen-binding fragment thereof in the manufacture of a reagent for diagnosing an FZD10-related disease or a predisposition for developing the disease.
  • the present invention provides a method for detecting an FZD10 inhibitor treatment responsive marker, comprising the step of detecting FZD10 protein in a sample as the responsive marker using the antibody of the present invention or an antigen-binding fragment thereof.
  • a method of including is provided.
  • the expression of FZD10 is specifically enhanced in certain types of cancer cells, and it has been clarified that the proliferation of such cancer cells is suppressed by FZD10 inhibitors (WO2005 / 004912, WO2006 / 013733, WO2007 / 148417). That is, responsiveness to FZD10 inhibitors can be predicted using FZD10 expression as an index.
  • an FZD10 inhibitor treatment responsive marker is an FZD10 protein in a sample isolated from a subject that is detected by binding to an antibody of the present invention or an antigen-binding fragment thereof, When the expression level is comparable or higher than the control level, the subject is shown to have a high therapeutic effect by the FZD10 inhibitor.
  • the control level is preferably a disease control level, i.e.
  • the present invention also provides an antibody of the present invention or an antigen-binding fragment thereof for use in screening a subject having a high therapeutic effect with an FZD10 inhibitor.
  • the present invention provides the use of an antibody of the present invention or an antigen-binding fragment thereof in the manufacture of a reagent for screening a subject with a high therapeutic effect by an FZD10 inhibitor.
  • the present invention further provides a method for detecting a drug efficacy marker of an FZD10 inhibitor, the method comprising the step of detecting FZD10 protein in a sample as the drug efficacy marker using the antibody of the present invention or an antigen-binding fragment thereof.
  • the expression of FZD10 is specifically enhanced in certain types of cancer cells, and it has been clarified that the proliferation of such cancer cells is suppressed by FZD10 inhibitors (WO2005 / 004912, WO2006 / 013733, WO2007 / 148417).
  • FZD10 inhibitors WO2005 / 004912, WO2006 / 013733, WO2007 / 148417.
  • cancerous tissue with such cancer cells can be reduced or killed by FZD10 inhibitors.
  • the efficacy of the FZD10 inhibitor in a subject having such cancer cells can be evaluated using the expression level of FZD10 as an index. If the expression level of FZD10 in a sample isolated from tissue that had FZD10 positive cancer cells is reduced compared to the sample isolated before treatment with the FZD10 inhibitor, the FZD10 inhibitor This is because FZD10-positive cancer cells can be considered to have decreased. Therefore, if the expression level of FZD10 can be specifically detected, it is useful as a drug efficacy marker for FZD10 inhibitors.
  • a FZD10 inhibitor drug efficacy marker is a sample in a sample isolated from a subject that has been administered an FZD10 inhibitor that is detected by binding to an antibody of the present invention or an antigen-binding fragment thereof.
  • FZD10 protein characterized in that when the expression level is low compared to the control level, the subject was shown to have a medicinal effect of the FZD10 inhibitor.
  • the control level is preferably the expression level of FZD10 protein in a sample isolated from the disease site of interest prior to drug administration.
  • the present invention also provides an antibody of the present invention or an antigen-binding fragment thereof for use in determining the efficacy after treatment with an FZD10 inhibitor.
  • the present invention provides the use of an antibody of the present invention or an antigen-binding fragment thereof in the manufacture of a reagent for determining drug efficacy after treatment with an FZD10 inhibitor.
  • kits for diagnosis of FZD10-related diseases for diagnosis of FZD10-related diseases, screening of subjects with high therapeutic effects with FZD10 inhibitors, or determination of drug efficacy after treatment with FZD10 inhibitors
  • the present invention relates to diagnosis of FZD10-related diseases, FZD10 inhibitors
  • the present invention provides a reagent or kit for screening a subject having a high therapeutic effect by, or for determining a drug effect after treatment with an FZD10 inhibitor.
  • these kits contain the antibody of the present invention or an antigen-binding fragment thereof as a detection reagent for FZD10 protein.
  • an antibody for a diagnostic reagent or kit of the present invention can be labeled with a fluorescent material, a luminescent material, or a radioisotope.
  • a fluorescent material e.g., a fluorescent material
  • a luminescent material e.g., a fluorescent material
  • a radioisotope e.g., a radioisotope
  • the kit can contain a combination of the antibody of the present invention or an antigen-binding fragment thereof and another marker detection reagent.
  • the kit can further include positive and negative control reagents for FZD10 and a secondary antibody for detecting the antibody of the present invention.
  • a culture section or tissue sample of a cell line known to be highly expressing FZD10 can serve as a useful positive control reagent.
  • tissue samples obtained from healthy subjects or non-cancerous tissues can serve as useful negative control reagents.
  • the secondary antibody for detecting the antibody of the present invention is preferably labeled with a fluorescent substance, a luminescent substance, a radioisotope, or an enzyme.
  • the kit of the present invention further includes buffers, diluents, filters, needles, syringes, and package inserts with instructions for use (eg, written, tape, CD-ROM, etc.) for commercial or user standpoints. May contain other materials desired. These reagents and the like can be held in a labeled container. Suitable containers include bottles, vials, and test tubes. The container can be formed from a variety of materials such as glass or plastic.
  • FZD10 forced expression cell line (FZD10 / DLD1) in which the FZD10 expression vector was introduced into DLD1, a human colon cancer cell line purchased from ATCC, and an empty vector introduced as a negative control (Mock / DLD1) was produced.
  • DLD1 a human colon cancer cell line purchased from ATCC
  • Mock / DLD1 an empty vector introduced as a negative control
  • SYO-1 a human synovial sarcoma cell line distributed by the National Cancer Center Rare Cancer Center, is 10% fetal bovine serum (FBS) and 1% at 37 ° C in a humidified atmosphere of 5% CO 2 Maintained in DMEM supplemented with penicillin / streptomycin.
  • TT a human esophageal cancer cell line purchased from JCRB, is DMEM / F12 (1: 5) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin / streptomycin at 37 ° C. in a humidified atmosphere of 5% CO 2. 1) maintained in.
  • H727 a human lung cancer cell line purchased from ATCC, was maintained in RPMI-1640 supplemented with 10% fetal bovine serum (FBS) and 1% penicillin / streptomycin at 37 ° C. in a humidified atmosphere of 5% CO 2 .
  • COLO201 a human colon cancer cell line purchased from ATCC, is 10% fetal bovine serum (FBS), 1% penicillin / streptomycin, 10 mM HEPES, 1 mM sodium pyruvate and 4.5 at 37 ° C. in a humidified atmosphere of 5% CO 2.
  • LoVo a human colon cancer cell line purchased from ATCC, was maintained in F12 supplemented with 20% fetal bovine serum (FBS) and 1% penicillin / streptomycin at 37 ° C. in a humidified atmosphere of CO 2 .
  • FBS fetal bovine serum
  • penicillin / streptomycin at 37 ° C. in a humidified atmosphere of CO 2 .
  • Immunohistochemical staining Formalin-fixed paraffin-sectioned FZD10 / DLD1, Mock / DLD1, SYO1, TT, H727, LoVo, COLO201 and clinical specimens were immersed in xylene three times for 3 minutes and then deparaffinized, and then twice in 100% ethanol for 1 minute , 90%, 70%, and 50% ethanol for 1 minute each to rehydrate.
  • Sections immersed in antigen activation solution pH9 (Nichirei Bioscience) for antigen activation treatment are FZD10 / DLD1, Mock / DLD1, 125 ° C for 30 seconds, SYO1, TT, H727, LoVo, COLO201, and 95 clinical specimens.
  • FZD10Ab (10A8H4G4), FZD10 (L164) pAb, FZD10 PolyclonalAb were appropriately reduced in a wet box, allowed to stand for 60 minutes, and then washed 3 times with Wash Buffer for 5 minutes.
  • An appropriate amount of Histofine Simple Stain MAX-PO was used as a secondary antibody in a wet box and allowed to stand for 30 minutes. Wash 3 times for 5 minutes with Wash Buffer. Color development reaction was performed with DAB substrate solution (Nichirei Bioscience). The slide section was immersed in hematoxylin (Dako) for 20 seconds and washed with running water.
  • Dehydration was performed by immersion in 50%, 70%, and 90% ethanol for 1 minute each and twice in 100% ethanol for 1 minute each. Finally, the sections were immersed in xylene twice for 3 minutes to be clarified, and mounted by Mount-Quick (DAIDO SANGYO).
  • RNeasy Mini kit QIAGEN
  • the frozen tissue piece was soaked in TRIzol Reagent (Invitrogen) and ground and extracted with chloroform. An approximately equal amount of 70% ethanol was added to the obtained extract, and total RNA was extracted using RNeasy Mini kit (QIAGEN).
  • CDNA was synthesized from total RNA using reverse transcriptase of SuperScript III Reverse Transcriptase (Invitrogen). PCR reaction was performed using KAPA SYBR FAST ABI Prism qPCR kit (KAPA Biosystems) using cDNA as a template.
  • the target gene for expression analysis was FZD10, and the housekeeping gene was GAPDH (glyceraldehyde-3-phosphate dehydrogenase).
  • FZD10 uses FZD10-11F: 5'-GTGTGCAGCCGTAGGTTAAAG-3 '(SEQ ID NO: 12), FZD10 R5: 5'-GACTGGGCAGGGATCTCATA-3' (SEQ ID NO: 13) primer set, GAPDH is GAPDH RT-PCR Fw: 5'-ACAACAGCCTCAAGATCATCAG-3 '(SEQ ID NO: 14), GAPDH RT-PCR Re: 5'-GGTCCACCACTGACACGTTG-3' (SEQ ID NO: 15) I did it.
  • the amount of PCR amplification product was monitored over time, and a Ct value was calculated by setting a threshold in a region where the PCR amplification product was exponentially amplified.
  • the concentration of the unknown sample was calculated by applying the Ct value to the calibration curve.
  • FCM Flow cytometry
  • Example 1 Production of anti-FZD10 monoclonal antibody (1) Acquisition of anti-FZD10 antibody-producing hybridoma using soluble FZD10 protein as an immunogen Since FZD10 is a membrane protein, it can be used to produce antibodies that react with immobilized membrane proteins. The extracellular region can be an immunogen. Since the extracellular region other than the N-terminus of FZD10 was very short and thought to be difficult to use as an antigen, the N-terminal extracellular region (1-161 amino acids) was selected as the immunogen, and the signal peptide Monoclonal antibodies were prepared using soluble FZD10 protein (21-161 amino acid region) (SEQ ID NO: 9) except for.
  • mice After adding 50 ⁇ g of the antigenic peptide to Freund Adjuvant, it was emulsified and injected for the first time by subcutaneous injection into Balb / c mice (Japan SLC Co., Ltd.). The second and subsequent immunizations were carried out by injecting subcutaneously the equivalent amount of 25 ⁇ g antigen peptide prepared in the same manner. Three days after the final immunization, spleen cells were aseptically prepared from mice, and cell fusion with mouse myeloma cells SP2 / 0 was performed by the polyethylene glycol method according to a conventional method.
  • Anti-FZD10 antibody was selected by first screening for an antibody having binding ability to FZD10 by ELISA using soluble FZD10 protein. The antibodies selected by ELISA were further selected by immunostaining. Selection by immunostaining was performed by immunohistochemical staining using FZD10 forced expression cell line FZD10 / DLD1 expressing full-length FZD10 protein (SEQ ID NO: 22). That is, FZD10 forced expression cell line FZD10 / DLD1 in which full-length FZD10 protein was forcibly expressed was sectioned with formalin-fixed paraffin and then immunostained to select hybridomas in which a strong reaction was observed.
  • FIG. 2 shows the results of immunohistochemical staining for the hybridoma clone 10A8H4G4 that was confirmed to produce a high level of FZD10-specific antibody among the tested hybridomas.
  • FIG. 2 shows the results of immunohistochemical staining for the hybridoma clone 10A8H4G4 that was confirmed to produce a high level of FZD10-specific antibody among the tested hybridomas.
  • FIG. 2 shows the results of immunohistochemical staining for the hybridoma clone 10A8H4G4 that was confirmed to produce a high level of FZD10-specific antibody among the tested hybridomas.
  • FIG. 2 shows the results of immunohistochemical staining for the hybridoma clone 10A8H4G4 that was confirmed to produce a high level of FZD10-specific antibody among the tested hybridomas.
  • the hybridoma clone 10A8H4G4 is useful as a hybridoma producing an antibody that specifically detects FZD10 in immunohistochemical staining.
  • staining was performed using commercially available antibodies (FZD10 (L164) Ab, FZD10 Polyclonal Ab), staining was observed in both FZD10 / DLD1 and Mock / DLD1 (a signal was detected in a high proportion of cells). Therefore, it was suspected that the detection results with these commercially available antibodies contained a false positive signal.
  • the anti-FZD10 antibody (10A8H4G4) of the present invention has an advantageous property that FZD10 can be detected with high specificity in the immunohistochemical staining method as compared with a commercially available anti-FZD10 antibody. Became clear.
  • This hybridoma clone 10A8H4G4 was selected to produce antibodies for further experiments.
  • the hybridoma clone 10A8H4G4 was mass-cultured, and the culture solution was collected after 2 to 3 weeks.
  • the antibody was purified from the culture using a protein A column (GE Healthcare, NJ).
  • the antibody of the present invention is also referred to as clone 10A8H4G4.
  • Example 2 Evaluation of specificity of anti-FZD10 monoclonal antibody
  • 10A8H4G4 was evaluated using various cell lines with different expression levels of FZD10. That is, the results of immunohistochemical staining and flow cytometry using 10A8H4G4 were compared, and it was examined whether staining corresponding to the expression level of FZD10 protein was observed.
  • CCLE cancer cell line encyclopedia
  • an mRNA expression analysis database using cell lines for each cancer type marked up-regulation is shown in lung cancer, esophageal cancer, and colorectal cancer (colon cancer). ( Figure 1). Therefore, cell lines derived from these cancer types were used for the following analysis.
  • endogenous FZD10 protein on the cell membrane of various human cancer tissue-derived cell lines was confirmed by flow cytometry using 10A8H4G4.
  • 10A8H4G4 in addition to human synovial sarcoma-derived SYO-1, which is known to express endogenous FZD10 protein, in human esophageal cancer-derived cell line TT and human lung cancer-derived cell line H727 A positive signal was detected by flow cytometry using 10A8H4G4.
  • no positive signal was detected by flow cytometry using 10A8H4G4 in the human colon cancer-derived cell line LoVo, which is known to be an FZD10 non-expressing cell line.
  • paraffin sections prepared from these cell lines were subjected to immunohistochemical staining using 10A8H4G4.
  • staining with 10A8H4G4 revealed staining in paraffin sections prepared from cell lines SYO-1, TT, and H727 that were confirmed to express FZD10. .
  • a positive signal was detected in a high proportion of cells in correlation with the flow cytometry results.
  • paraffin sections prepared from LoVo which is a non-cell line of FZD10, were hardly stained, and positive signals were hardly detected.
  • the anti-FZD10 antibody (10A8H4G4) of the present invention is a useful tool capable of detecting FZD10 protein in a sample with high specificity in both flow cytometry and immunohistochemical staining. .
  • Example 3 Detection of FZD10 protein in tumor transplantation models and clinical specimens Specificity in immunohistochemical staining of the anti-FZD10 antibody (10A8H4G4) of the present invention was determined using a paraffin section prepared from a mouse Xenograft tumor of a cancer cell line, and Evaluation was performed using clinical specimens of lung cancer. Paraffin sections prepared from mouse Xenograft tumors of SYO-1 and COLO201 cell lines and expression of FZD10 protein and RNA in tumor and non-tumor areas of lung cancer clinical specimens 1-6 by immunohistochemical staining and RealTime-PCR, respectively The detection results are shown in FIG. As shown in FIG.
  • lung cancer clinical specimens 5 in which the expression of tumor region-specific FZD10 RNA was confirmed cells stained with 10A8H4G4, that is, FZD10-positive cells were detected, but tumor region-specific FZD10 RNA expression was detected.
  • the other lung cancer specimens that were not confirmed were hardly stained and few FZD10 positive cells were detected. This result indicates that 10A8H4G4 can specifically detect FZD10 protein even in clinical specimens.
  • Example 4 Analysis of amino acid sequence of variable region of anti-FZD10 monoclonal antibody
  • the amino acid sequence of the variable region of the anti-FZD10 antibody (10A8H4G4) of the present invention was analyzed.
  • Total RNA was extracted from hybridoma 10A8H4G4 using RNeasy mini kit (QIAGEN).
  • CDNA was synthesized from total RNA using Super Script II Reverse Transcriptase (Invitrogen). Primers for cDNA synthesis are as follows.
  • MIGCUniRv for heavy chain 3 'primer 5'-CTGGGAAGGTGTGCACAC-3 '(SEQ ID NO: 16)
  • MIGKRv2 light chain 3 'primer 5'-GTTGTTCAAGAAGCACACGAC-3 '(SEQ ID NO: 17)
  • a 5 'RACE System for Rapid Amplification of cDNA Ends (Invitrogen) is used to add a dC polymer to the cDNA end, and Platinum Taq DNA Polymerase High Fidelity (Invitrogen) is used to encode a polynucleotide encoding the variable region of a monoclonal antibody. Amplified. Primers for amplification are as follows.
  • Base “I” in the primer sequence indicates inosine.
  • 5 'RACE Abridged Anchor Primer for heavy chain 5' and light chain 5 'primers 5'-GGCCACGCGTCGACTAGTACGGGIIGGGIIGGGIIG-3 '(SEQ ID NO: 18), MIGCUniRv2 for heavy chain 3 'primer: 5′-TGGACAGGGATCCAGAGTTCC-3 ′ (SEQ ID NO: 19), as well as light chain 3 ′ primer mIGKNesRv2: 5′-CAGATGTTAACTGCTCACTGGATGG-3 ′ (SEQ ID NO: 20).
  • the PCR product was cloned into pGEM-T Easy Vector (Promega). The insert fragment region was sequenced, and the nucleic acid sequence of the variable region of 10A8H4G4 (excluding the signal sequence) was determined.
  • amino acid sequence and nucleic acid sequence of the heavy chain variable region and light chain variable region of the mouse monoclonal antibody were determined as follows. 10A8H4G4, heavy chain variable region amino acid sequence (excluding signal sequence): QVTLKESGPGILQPSQTLSLTCSFSGFSLSTSGLGVSWIRQPSGKGLEWLAHIYWDDDKRYNPSLKSRLTISKDTSSNQVFLKITSVDTADTATYYCARRAYYGNYYALDYWGQGTSVTVSS (SEQ ID NO: 7 encoded by the nucleic acid sequence shown in SEQ ID NO: 10) 10A8H4G4, heavy chain variable region nucleic acid sequence: 5'-CAGGTTACTCTGAAAGAGTCTGGCCCTGGGATATTGCAGCCCTCCCAGACCCTCAGTCTGACTTGTTCTTTCTCTGGGTTTTCACTGAGCACTTCTGGTCTGGGTGTGAGCTGGATTCGTCAGCCTTCAGGAAAGGGTCTGGAGTGGCTGGCACACATTTACTGGGATGATGACAAGCG
  • the CDR sequence of the antibody of the present invention (10A8H4G4) determined by Kabat's definition is as follows.
  • Heavy chain CDR1 (CDR-H1) TSGLGVS (SEQ ID NO: 1);
  • Heavy chain CDR2 (CDR-H2) HIYWDDDKRYNPSLKS (SEQ ID NO: 2);
  • Light chain CDR2 (CDR-L2) WASTRKS (SEQ ID NO: 5); and
  • Light chain CDR3 (CDR-L3) QNDYSYPVT (SEQ ID NO: 6)
  • the present invention has succeeded in producing an anti-FZD10 antibody capable of detecting FZD10 protein in a sample isolated from a subject such as a clinical specimen with high specificity.
  • FZD10 antibody of the present invention FZD10 protein in a sample can be detected with high sensitivity and low background. Therefore, the antibody of the present invention is useful for diagnosis of FZD10-related diseases such as cancers that express FZD10.
  • the antibody of the present invention can detect FZD10 according to the expression level of FZD10 in a sample, screening of subjects with high therapeutic effects by FZD10 inhibitors (pre-treatment diagnosis) and FZD10 inhibition in subjects It is also useful in the determination of drug efficacy after treatment with agents (post-treatment diagnosis).

Abstract

L'invention concerne un anticorps monoclonal anti-FZD10. En outre, l'invention concerne un procédé de diagnostic des maladies associées au FZD10 mettant en œuvre cet anticorps, un procédé de détection de protéine FZD10, un procédé jugeant de l'efficacité de médicaments après traitement au moyen d'un inhibiteur de FZD10, un procédé de dépistage de sujets présentant un important effet thérapeutique au moyen de l'inhibiteur de FZD10, et un réactif de diagnostic contenant cet anticorps.
PCT/JP2017/036081 2016-10-06 2017-10-04 Anticorps monoclonal anti-fzd10, et application de celui-ci WO2018066585A1 (fr)

Priority Applications (9)

Application Number Priority Date Filing Date Title
KR1020197012936A KR102531006B1 (ko) 2016-10-06 2017-10-04 Fzd10에 대한 모노클로널 항체 및 이의 사용
US16/336,846 US11079386B2 (en) 2016-10-06 2017-10-04 Monoclonal antibody against FZD10 and use thereof
EP17858422.3A EP3524677B1 (fr) 2016-10-06 2017-10-04 Anticorps monoclonal anti-fzd10, et application de celui-ci
CA3038789A CA3038789A1 (fr) 2016-10-06 2017-10-04 Anticorps monoclonal anti-fzd10, et application de celui-ci
BR112019006422A BR112019006422A2 (pt) 2016-10-06 2017-10-04 anticorpo monoclonal contra fzd10 e utilização do mesmo
CN201780075513.0A CN110036110B (zh) 2016-10-06 2017-10-04 针对fzd10的单克隆抗体及其用途
RU2019112825A RU2765431C2 (ru) 2016-10-06 2017-10-04 Моноклональное антитело против fzd10, его использование
JP2018543931A JP7039039B2 (ja) 2016-10-06 2017-10-04 Fzd10に対するモノクローナル抗体およびその使用
JP2022004142A JP2022068145A (ja) 2016-10-06 2022-01-14 Fzd10に対するモノクローナル抗体およびその使用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2016-197881 2016-10-06
JP2016197881 2016-10-06

Publications (1)

Publication Number Publication Date
WO2018066585A1 true WO2018066585A1 (fr) 2018-04-12

Family

ID=61831895

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2017/036081 WO2018066585A1 (fr) 2016-10-06 2017-10-04 Anticorps monoclonal anti-fzd10, et application de celui-ci

Country Status (10)

Country Link
US (1) US11079386B2 (fr)
EP (1) EP3524677B1 (fr)
JP (2) JP7039039B2 (fr)
KR (1) KR102531006B1 (fr)
CN (1) CN110036110B (fr)
BR (1) BR112019006422A2 (fr)
CA (1) CA3038789A1 (fr)
RU (1) RU2765431C2 (fr)
TW (1) TWI762516B (fr)
WO (1) WO2018066585A1 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI762516B (zh) * 2016-10-06 2022-05-01 日商腫瘤療法 科學股份有限公司 針對fzd10之單株抗體及其用途
CN110835372B (zh) * 2019-11-06 2020-12-01 上海健康医学院 一种靶向Frizzled7单克隆抗体及其制备方法与应用

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
WO1993016185A2 (fr) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Proteine de liaison biosynthetique pour marqueur de cancer
WO1994011523A2 (fr) 1992-11-13 1994-05-26 Idec Pharmaceuticals Corporation Sequences kozak consensus totalement alterees destinees a l'expression chez les mammiferes
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
US5641870A (en) 1995-04-20 1997-06-24 Genentech, Inc. Low pH hydrophobic interaction chromatography for antibody purification
WO2004020668A2 (fr) 2002-08-30 2004-03-11 Oncotherapy Science, Inc. Methode de traitement du synovialome
WO2005004912A1 (fr) 2003-07-11 2005-01-20 Oncotherapy Science, Inc. Methode de traitement du synovialome
WO2006013733A1 (fr) 2004-08-05 2006-02-09 The University Of Tokyo PROCÉDÉ SERVANT À TRAITER LE SARCOME SYNOVIAL UTILISANT UN ARNsi POUR LE FZD10
WO2007053577A2 (fr) 2005-10-31 2007-05-10 Oncomed Pharmaceuticals, Inc. Compositions et procedes pour diagnostiquer et traiter un cancer
WO2007148417A1 (fr) 2006-06-21 2007-12-27 Oncotherapy Science, Inc. Anticorps monoclonaux de ciblage tumoral dirigés contre la fzd10 et leurs utilisations
WO2013157410A1 (fr) * 2012-04-17 2013-10-24 Hoya株式会社 Peptide de liaison de fzd10

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9414580D0 (en) 1994-07-19 1994-09-07 Cancer Res Campaign Tech Materials and methods relating to the diagnosis of synovial sarcomas
EP1272224A4 (fr) 2000-03-31 2004-09-29 Gene Logic Inc Profils d'expression genique dans un tissu oesophagien
AU2001284160A1 (en) 2000-08-19 2002-03-04 Axordia Limited Modulation of stem cell differentiation
WO2002055705A2 (fr) 2001-01-11 2002-07-18 Curagen Corp Proteines et acides nucleiques codant ces proteines
US6964849B2 (en) 2001-01-11 2005-11-15 Curagen Corporation Proteins and nucleic acids encoding same
WO2002086443A2 (fr) 2001-04-18 2002-10-31 Protein Design Labs, Inc Procedes de diagnostic du cancer du poumon, compositions et procedes de criblage de modulateurs du cancer du poumon
US7682607B2 (en) 2001-05-01 2010-03-23 The Regents Of The University Of California Wnt and frizzled receptors as targets for immunotherapy in head and neck squamous cell carcinomas
US20030044409A1 (en) 2001-05-01 2003-03-06 Carson Dennis A. Immunologic compositions and methods for studying and treating cancers expressing frizzled antigens
WO2003004045A2 (fr) 2001-07-05 2003-01-16 Xenon Genetics, Inc. Procedes permettant d'identifier des agents therapeutiques pour traiter des maladies impliquant des polypeptides wnt et des recepteurs wnt
US7803370B2 (en) * 2002-08-30 2010-09-28 Oncotherapy Science, Inc. Method for treating synovial sarcoma
TW200413725A (en) 2002-09-30 2004-08-01 Oncotherapy Science Inc Method for diagnosing non-small cell lung cancers
US20060024692A1 (en) 2002-09-30 2006-02-02 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
US20040247593A1 (en) 2002-10-04 2004-12-09 Regents Of The University Of California, Methods for treating cancer by inhibiting Wnt signaling
EP2295601A1 (fr) 2005-02-10 2011-03-16 Oncotherapy Science, Inc. Procédé de diagnostic de cancer de la vessie
SG190568A1 (en) * 2008-09-26 2013-06-28 Oncomed Pharm Inc Frizzled-binding agents and uses thereof
US20130295106A1 (en) * 2008-09-26 2013-11-07 Oncomed Pharmaceuticals, Inc. Frizzled-Binding Agents And Uses Thereof
EP3783028A1 (fr) * 2011-08-12 2021-02-24 Omeros Corporation Anticorps monoclonaux anti-fzd10 et leurs procédés d'utilisation
TWI762516B (zh) * 2016-10-06 2022-05-01 日商腫瘤療法 科學股份有限公司 針對fzd10之單株抗體及其用途

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
WO1993016185A2 (fr) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Proteine de liaison biosynthetique pour marqueur de cancer
WO1994011523A2 (fr) 1992-11-13 1994-05-26 Idec Pharmaceuticals Corporation Sequences kozak consensus totalement alterees destinees a l'expression chez les mammiferes
US5641870A (en) 1995-04-20 1997-06-24 Genentech, Inc. Low pH hydrophobic interaction chromatography for antibody purification
WO2004020668A2 (fr) 2002-08-30 2004-03-11 Oncotherapy Science, Inc. Methode de traitement du synovialome
WO2005004912A1 (fr) 2003-07-11 2005-01-20 Oncotherapy Science, Inc. Methode de traitement du synovialome
JP2007526891A (ja) * 2003-07-11 2007-09-20 オンコセラピー・サイエンス株式会社 滑膜肉腫の治療方法
WO2006013733A1 (fr) 2004-08-05 2006-02-09 The University Of Tokyo PROCÉDÉ SERVANT À TRAITER LE SARCOME SYNOVIAL UTILISANT UN ARNsi POUR LE FZD10
WO2007053577A2 (fr) 2005-10-31 2007-05-10 Oncomed Pharmaceuticals, Inc. Compositions et procedes pour diagnostiquer et traiter un cancer
WO2007148417A1 (fr) 2006-06-21 2007-12-27 Oncotherapy Science, Inc. Anticorps monoclonaux de ciblage tumoral dirigés contre la fzd10 et leurs utilisations
JP2009541204A (ja) * 2006-06-21 2009-11-26 オンコセラピー・サイエンス株式会社 Fzd10に対する腫瘍標的化モノクローナル抗体とその使用
WO2013157410A1 (fr) * 2012-04-17 2013-10-24 Hoya株式会社 Peptide de liaison de fzd10

Non-Patent Citations (37)

* Cited by examiner, † Cited by third party
Title
"Antibodies A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY
"Antibodies: A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY
"Genbank", Database accession no. NM 007197.3
"Strategies for Protein Purification and Characterization: A Laboratory Course Manual", 1996, COLD SPRING HARBOR LABORATORY PRESS
BEAUCAGE SL; IYER RP, TETRAHEDRON, vol. 48, 1992, pages 2223 - 311
BETTER ET AL., SCIENCE, vol. 240, 1988, pages 1041 - 3
BRENNAN ET AL., SCIENCE, vol. 229, 1985, pages 81
BRODEUR ET AL.: "Monoclonal Antibody Production Techniques and Applications", 1987, MARCEL DEKKER, INC., pages: 51 - 63
CARTER ET AL., BIO/TECHNOLOGY, vol. 10, 1992, pages 163 - 167
CHARLES E. DANN ET AL., NATURE, vol. 412, 2001, pages 86 - 90
CHOTHIA ET AL., J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CLACKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628
EBERT, K.M. ET AL., BIO/TECHNOLOGY, vol. 12, 1994, pages 699 - 702
FASBE J., vol. 6, 1992, pages 2422 - 7
GODING: "Monoclonal Antibodies: Principles and Practice", 1986, ACADEMIC PRESS, pages: 59 - 103
H. TERASAKI ET AL., INT. J. MOL. MED., vol. 9, 2002, pages 107 - 112
HUSE ET AL., SCIENCE, vol. 246, 1989, pages 1275 - 1281
JONES ET AL., NATURE, vol. 321, 1986, pages 522 - 5
KABAT E. A. ET AL.: "Sequence of Proteins of Immunological Interest", 1991
KOHLER ET AL., NATURE, vol. 256, 1975, pages 495
KOZBOR, J. IMMUNOL., vol. 133, 1984, pages 300 1
LEI, S.P. ET AL., J. BACTERIOL., vol. 169, 1987, pages 4379 - 83
MARKS ET AL., BIOTECHNOLOGY, vol. 10, 1992, pages 779 - 783
MARKS ET AL., J MOL BIOL, vol. 222, 1991, pages 581 - 597
MATTHES ET AL., EMBO J, vol. 3, 1984, pages 801 - 5
MCCAFFERTY ET AL., NATURE, vol. 348, 1990, pages 552 - 554
MIZUSHIMA ET AL., NUCLEIC ACIDS RES., vol. 18, 1990, pages 5322
MORIMOTO ET AL., JOURNAL OF BIOCHEMICAL AND BIOPHYSICAL METHODS, vol. 24, 1992, pages 107 - 117
MULLIGAN ET AL., NATURE, vol. 277, 1979, pages 108 - 14
MUNSON ET AL., ANAL. BIOCHEM., vol. 107, 1980, pages 220
PLUCKTHUN, IMMUNOL. REVS., vol. 130, 1992, pages 151 - 188
S. NAGAYAMA ET AL., ONCOGENE, vol. 24, 2005, pages 6201 - 6212
SKERRA ET AL., CURR. OPINION IN IMMUNOL., vol. 5, 1993, pages 256 - 262
VANDAMME, A. M. ET AL., EUR. J. BIOCHEM., vol. 192, 1990, pages 767 - 75
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546
WARD ET AL., NATURE, vol. 341, pages 544 - 6
WATERHOUSE ET AL., NUC. ACIDS. RES., vol. 21, 1993, pages 2265 - 2266

Also Published As

Publication number Publication date
CA3038789A1 (fr) 2018-04-12
EP3524677B1 (fr) 2023-05-31
RU2765431C2 (ru) 2022-01-31
TWI762516B (zh) 2022-05-01
EP3524677A4 (fr) 2020-03-11
KR20190059318A (ko) 2019-05-30
EP3524677A1 (fr) 2019-08-14
CN110036110B (zh) 2023-02-03
JPWO2018066585A1 (ja) 2019-07-25
BR112019006422A2 (pt) 2019-06-25
RU2019112825A (ru) 2020-11-09
TW201827465A (zh) 2018-08-01
JP2022068145A (ja) 2022-05-09
JP7039039B2 (ja) 2022-03-22
US20200174002A1 (en) 2020-06-04
CN110036110A (zh) 2019-07-19
US11079386B2 (en) 2021-08-03
KR102531006B1 (ko) 2023-05-09
RU2019112825A3 (fr) 2021-02-26

Similar Documents

Publication Publication Date Title
US10775383B2 (en) PD-L1 antibodies and uses thereof
CN107011439B (zh) 抗叶酸受体α抗体及其应用
JP2022068145A (ja) Fzd10に対するモノクローナル抗体およびその使用
JP2022526131A (ja) 肝細胞癌診断用バイオマーカーセレブロンと、これに特異的な新規なモノクローナル抗体
KR20180120716A (ko) 대상체가 췌관 선암을 앓을 위험을 평가하기 위한 초기 및 비 침습적 방법 및 이러한 질환의 치료 방법
JP7094557B2 (ja) Melkに対するモノクローナル抗体およびその使用
WO2021030450A1 (fr) Nouvelles compositions d'anticorps anti-lox -1, dosage de neutralisation de lox1 et méthodes de traitement les utilisant

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17858422

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2018543931

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3038789

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112019006422

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20197012936

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017858422

Country of ref document: EP

Effective date: 20190506

ENP Entry into the national phase

Ref document number: 112019006422

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20190329